Download Document 39209

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Butyric acid wikipedia , lookup

Ribosomally synthesized and post-translationally modified peptides wikipedia , lookup

Nicotinamide adenine dinucleotide wikipedia , lookup

Microbial metabolism wikipedia , lookup

Mitogen-activated protein kinase wikipedia , lookup

Oxidative phosphorylation wikipedia , lookup

NADH:ubiquinone oxidoreductase (H+-translocating) wikipedia , lookup

Glycolysis wikipedia , lookup

Fatty acid synthesis wikipedia , lookup

Citric acid cycle wikipedia , lookup

Artificial gene synthesis wikipedia , lookup

Paracrine signalling wikipedia , lookup

Evolution of metal ions in biological systems wikipedia , lookup

Metalloprotein wikipedia , lookup

Hepoxilin wikipedia , lookup

Biochemical cascade wikipedia , lookup

Catalytic triad wikipedia , lookup

Metabolism wikipedia , lookup

Biochemistry wikipedia , lookup

Enzyme inhibitor wikipedia , lookup

Biosynthesis of doxorubicin wikipedia , lookup

Enzyme wikipedia , lookup

Biosynthesis wikipedia , lookup

Amino acid synthesis wikipedia , lookup

Transcript
Int. J. Pharm. Sci. Rev. Res., 25(2), Mar – Apr 2014; Article No. 42, Pages: 221-230
ISSN 0976 – 044X
Research Article
Exploration of Lysine Pathway for Developing Newer Anti-Microbial Analogs through
Enzyme Inhibition Approach
*
Mayura Kale , Mohammad Sayeed Shaikh
Department of Pharmaceutical Chemistry, Government College of Pharmacy, Aurangabad, Maharashtra, India.
*Corresponding author’s E-mail:
Accepted on: 05-02-2014; Finalized on: 31-03-2014.
ABSTRACT
Recently, a renewed interest towards development of new antibacterial agents has been created due to emergence of newer
pathogenic bacterial strains showing high resistance to such agents. There has also been a decline in research by medical and
pharmaceutical companies in last decade which has caused a shortfall in developing newer agents to fight present threat of drug
resistance. Hence, there is continuous need to develop newer antibiotics that interact with essential mechanisms in bacteria.
Recently, the enzymes responsible for biosynthesis of the essential amino acid lysine in plants, bacteria and fungi have been
targeted and it has augmented interest to develop novel antibiotic compounds and to enhance lysine yields in over-producing
organisms. Diaminopimelate (DAP) pathway in bacteria for biosynthesis of lysine and its immediate precursor meso-DAP have been
represented as novel targets, both of which play a major role in cross-linking of peptidoglycan layer of microbial cell wall. Lysine is a
constituent in gram-positive bacteria while gram-negative bacteria contain meso-DAP. Substrate-based inhibitors of enzymes in the
DAP pathway have been reviewed and inhibitors that allow better understanding of the enzymology of the targets and provide
insight for the design of new inhibitors have been discussed in this article. Synthetic enzyme inhibitors of the DAP pathway with
appropriate substrate-based analog have been found to be more effective against resistant bacterial strains and less toxic to
mammals. The enzymes involved in this pathway may be viable targets and shall be supportive to develop novel antimicrobial drugs.
Keywords: Bacterial resistance, Diaminopimelic acid, Diaminopimelate decarboxylase, Enzyme analogue, L-lysine.
INTRODUCTION
A
ntibiotics are the compounds that are literally
‘against life’ and are typically antibacterial drugs,
interfering with structure or process that is
essential for growth or survival of microorganisms with
least harm to the mammals. As we live in an era when
antibiotic resistance to microorganisms has spread at an
alarming rate, there is a constant need to develop newer
antibacterials which shall combat with the bacterial
survival strategies. However, clinically significant
resistance develops in periods of few months to years.
For penicillin, the resistance began to be noted within
two years of its introduction in the mid 1940s.1-3 The
recent emergence of mutated bacterial strains that are
resistant to currently available antibiotics has resulted in
renewed interest in the search for novel antibacterial
compounds. Such compounds should be targeted toward
proteins that are essential for bacterial viability but are
not present in mammals.4,5 The diaminopimelic acid and
lysine biosynthesis meets both of these criteria,
presenting multiple targets for novel antimicrobial
agents6,7. Lysine has been known to be an essential amino
acid required in protein synthesis and a constituent of the
peptidoglycan layer of cell walls in gram positive bacteria.
The lysine biosynthesis also produces D,L-diaminopimelic
acid (meso-DAP), which is also a component of the
peptidoglycan layer of gram negative bacteria and
mycobacterial cell walls. This review describes several
recent advancements in structure-based drug discovery in
the antimicrobial drugs which shall be useful to generate
new promising future drug candidates.8-10 Peptidoglycan
layer consists of a beta-1,4-linked polysaccharide of
alternating N-acetylglucosamine (NAG) and Nacetylmuramic acid (NAM) sugar units as building blocks
of cell wall. Attached to the lactyl side chain of NAM unit
is a pentapeptide (muramyl residues) side chain of
general structure L-Ala-g-D-Glu-X-D-Ala-D-Ala, where X is
either L–Lysine or meso-DAP.11-13 Formation of the cross
links makes the bacterial cell wall resistant to lysis.
Compounds which inhibit lysine or DAP biosynthesis could
therefore be very effective antibiotics, if targeted towards
cell wall biosynthesis. The enzymes which catalyze the
synthesis of L-lysine in plants and bacteria have attracted
interest from two directions; firstly from those interested
in inhibiting lysine biosynthesis as a strategy for the
development of novel antibiotic or herbicidal compounds
and secondly to enhance lysine yields in over-producing
organisms.14,15 Many peptidoglycan monomers, including
the potent toxin from B. pertussis and N. gonorrhoeae,
and similar DAP containing peptides, possess a range of
biological effects such as cytotoxicity, antitumor activities,
angiotensin converting enzyme (ACE) inhibitor,
immunostimulant,
and
sleep-inducing
biological
16-18
activities.
Lysine biosynthetic pathway in fungi
This is called as α-aminoadepate (AAA) pathway is a
biochemical pathway for the synthesis of the amino acid
L-lysine. In the eukaryotes, this pathway is unique to the
higher fungi (containing chitin in cell walls) and
euglenoids.19-22 It has also been reported in bacteria of
the genus thermus, like T. thermophilus, T. aquaticus
International Journal of Pharmaceutical Sciences Review and Research
Available online at www.globalresearchonline.net
221
Int. J. Pharm. Sci. Rev. Res., 25(2), Mar – Apr 2014; Article No. 42, Pages: 221-230
contain lysJ, genes that is essential for lysine
biosynthesis.23 L-lysine is the only the essential amino
acid that has two distinct biosynthetic pathways; the DAP
pathway in plants and bacteria and the AAA pathway in
24
euglenoids and higher fungi. The AAA pathway is unique
to fungi and is thus can be a potential target for the
rational design of antifungal drugs.25,26
A key step in biosynthesis of lysine AAA biosynthetic
pathway in fungus like Saccharomyces cerevisiae is
enzymatic reduction of R-aminoadipate to the
semialdehyde which requires two gene products, Lys2
and Lys5. Here, the gene Lys5 is a specific
posttranslational modification catalyst. The AAA pathway
utilizes α-ketoglutarate and acetyl CoA (AcCoA) serving as
the precursor for L-lysine. Extensive genetic, enzymatic
and regulatory studies of the lysine biosynthesis are being
carried out in this fungal species.27 Seven enzymes and
more than twelve non-linked genes are responsible for
the biosynthesis of lysine in S. cerevisiae. Steps
comprising the first half of the pathway leading to αaminoadipate take place in the mitochondria with the
exception being the synthesis of homocitrate in the
nucleus and second half of pathway converting αaminoadipate to l-lysine are carried out in the cytoplasm.
The first half of the AAA pathway shares many similarities
with the tricarboxylic acid (TCA) cycle. The pathway
(Figure 2). is initiated by the homocitrate synthase (HCS)
enzyme catalyzed condensation of acetyl CoA and αketoglutarate to give the enzyme bound intermediate
homocitryl CoA, which undergoes hydrolysis by the same
enzyme to give homocitrate as the product first
enzymatic reaction. Homoaconitase (HAc) catalyzes the
inter conversion of homocitrate and homoisocitrate
through the intermediate homoaconitate.28 The
homoisocitrate is then oxidatively decarboxylated by the
pyridine nucleotide-linked homoisocitrate dehydrogenase
(HIDH) to give α-ketoadipate. The next step is carried out
by
pyridoxal
5′-phosphate
(PLP)-dependent
aminotransferase (AAT) by using L-glutamate as the
amino donor to form α-Aminoadipate. the conversion of
α-aminoadipate (AAA) to lysine, in T. thermophilus is
similar to the latter portion of arginine biosynthesis. As
the first half of the pathway takes place in the
mitochondrion, the aminotransferase is being present in
both mitochondrion and cytoplasm. In the cytoplasm,
AAA is then reduced to α-aminoadipate-δ-semialdehyde
(AAS) via AAA reductase (AAR). This enzyme has to be
activated by a phosphopantetheinyl transferase (PPT).
Once the α-aminoadipate-δ-semialdehyde (AAS) is
formed, it is combined with glutamate and by this the
imine is reduced by NADPH to L–saccharopine, reaction
being catalyzed by saccharopine reductase (SR). Finally,
saccharopine dehydrogenase (SDH) catalyzes the
oxidative deamination of saccharopine to give L29,30
lysine.
AAA pathway in fungi is of significance as many fungal
alkaloids synthesize lysine as a structural element or
biosynthetic precursor. In addition, several AAA pathway
ISSN 0976 – 044X
intermediates are used to generate secondary
metabolites such as AAA as an essential precursor for the
synthesis of ACV δ-(L-α-aminoadipoyl)-L-cysteinyl-Dvaline) tripeptide which is further utilized as the biological
precursor of penicillin. Systemic fungal infections are the
most difficult infectious diseases in mammals to treat and
are life-threatening for individuals who are immunesuppressed, including AIDS, autoimmune diseases, and
chemotherapy and transplant surgery. Selective inhibition
of the enzymes of this pathway by appropriate substrate
analog to develop new antifungal drugs that are more
effective and less toxic and the enzymes involved in this
pathway may be viable targets for selective antifungal
agents.31
Figure 1: Lysine AAA biosynthetic pathway in fungi
Lysine biosynthetic pathway in bacteria
The synthesis of meso-DAP and lysine begins with the
phosphorylation
of
L-aspartate
to
form
Laspartylphosphate catalyzed by aspartate kinase. Both
E.coli and B.subtilis genomes encoding show three
aspartokinase isozymes, required for different
biosynthetic pathways starting from aspartate. E.coli has
two
bifunctional
aspartokinase/homo-serine
dehydrogenases, ThrA and MetL, and one monofunctional
aspartokinase LysC, which are involved in the threonine,
methionine and lysine synthesis. Transcription of the
aspartokinase genes in E.coli is regulated by appropriate
concentrations of the corresponding amino acids. In
addition, ThrA and LysC are feedback inhibited by
threonine and lysine; respectively.32,33 Aspartate
semialdehyde
dehydrogenase
converts
the
Laspartylphosphate to aspartate semialdehyde. The first
two steps of the DAP pathway is catalyzed by
aspartokinase
and
aspartate
semialdehyde
dehydrogenase (ASADH) are common for the biosynthesis
of amino acids of the aspartate family, like lysine,
threonine and methionine.34 Dihydrodipicolinate synthase
catalyses the condensation of pyruvate (PYR) and
aspartate semialdehyde (ASA) to form 4-hydroxy-2,3,4,5tetrahydro-L,L-dipicolinic acid (HTPA). This enzyme
belongs to the family of lyases, specifically the
35,36
hydrolyases, which cleaves carbon-oxygen bonds.
The
13
studies using C-labelled pyruvate demonstrate that the
International Journal of Pharmaceutical Sciences Review and Research
Available online at www.globalresearchonline.net
222
Int. J. Pharm. Sci. Rev. Res., 25(2), Mar – Apr 2014; Article No. 42, Pages: 221-230
product is the unstable heterocycle HTPA. Rapid
decomposition of the 13C-NMR signals of HTPA following
its production indicates that formation of Ldihydrodipicolinate (DHDP) occurs via a nonenzymatic
step. Dihydrodipicolinate reductase (DHDPR) catalyses
the pyridine nucleotide-dependent reduction of DHDP to
form L-2,3,4,5,-tetrahydrodipicolinate (THDP). THDP and
DHDP is synthesized from aspartate semialdehyde by the
products of the dapA and dapB genes (Figure 3).
However, the metabolic pathway then diverges into four
sub-pathways depending on the species, namely the
succinylase,
acetylase,
dehydrogenase
and
aminotransferase pathways from tetrahydrodipicolinate
are known in bacteria. The presence of multiple
biosynthetic pathways is probably a result of the
37
importance of DAP and lysine to bacterial survival . The
most common of the alternative metabolic routes is the
succinylase pathway, which is inherent to many bacterial
species including E.coli This sub-pathway begins with the
conversion of tetrahydrodipicolinate to N-succinyl-L-2amino-6-ketopimelate (NSAKP) catalyzed by 2,3,4,5tetrahydropyridine-2-carboxylate N-succinyltransferase
(dapD, THPC-NST),] NSAKP is then converted to Nsuccinyl-L,L-2,6,-diaminopimelate
(NSDAP)
by
Nsuccinyldiaminopimelate
aminotransferase
(DapC,
NSDAP-AT). Subsequently it is desuccinylated by
succinyldiaminopimelate desuccinylase (dapE,SDAP-DS)
ISSN 0976 – 044X
to form L,L-2,6-diaminopimelate (LL-DAP). As for the
succinylase pathway, the acetylase pathway involves four
enzymatic steps, but incorporates N-acetyl groups rather
than N-succinyl moieties. This pathway is common to
several Bacillus species, including B. subtilis and B.
anthracis.38 The succinylase pathway is utilized by all
gram negative and many gram positive bacteria, while the
acetylase pathway appears to be limited to certain
Bacillus species. There are one additional sub-pathways
39
that are less common to bacteria. Accordingly after the
formation of L,L-DAP, which is common in these pathway,
the enzyme L,L-Diaminopimelate epimerase (DAPE, dapF)
catalyzes the epimerization of L,L-DAP to form meso-DAP.
The
dehydrogenase
pathway,
Diaminopimelate
dehydrogenase (DAPDH) catalyzes the direct conversion
of tetrahydrodipicolinate to meso-DAP in the
dehydrogenase pathway of lysine biosynthesis. All these
alternative pathways then converge to utilize the same
enzyme for the final step of lysine biosynthesis, namely
diaminopimelate decarboxylase (DAPDC, LysA) which
catalyses the decarboxylation of meso-DAP to yield lysine
and carbon dioxide. This step is important for the overall
regulation of the lysine biosynthesis. Comparative
analysis of genes and regulatory elements Identify the
lysine-specific RNA element, named the LYS element, in
the regulatory regions of bacterial genes involved in
biosynthesis and transport of lysine.
L-aspartyl-phosphate
ASADH (asd)
L-asparted-semialdehyde
DHDPS (dapA)
(Met, Thr, Ile)
L-2,3-dihydrodipicolinate
DHDPR (dapB)
L-tetrahydrodipicolinate
Dehydrogenase pathway
Acetylase pathway
THDP-NAT
N-acetyl-L-amino- 6-ketopimilate
Succinylase pathway
THPC-NST(dap D)
N-succinyl-L-amino-6-ketopimilate
ATA
DAPDH
(ddh)
NSDAP-AT(dap C)
N-acetyl-L-L-6-diaminopimelate
N-succinyl-L-L-6-diaminopimilate
NAD-DAC
SDAP-DS(dap E)
L-L-diaminopimilate
DAPE (dap F)
meso-diaminopimilate
DAPDC (LysA)
L-lysine
Figure 2: Enzymes of the lysine biosynthetic pathway in bacteria
International Journal of Pharmaceutical Sciences Review and Research
Available online at www.globalresearchonline.net
223
Int. J. Pharm. Sci. Rev. Res., 25(2), Mar – Apr 2014; Article No. 42, Pages: 221-230
The LYS element includes regions of lysine-constitutive
mutations previously identified in E.coli and B. subtilis.
The lysine biosynthetic pathway has a special interest for
pharmacology, since the absence of DAP in mammalian
cells allows for the use of the lysine biosynthetic genes as
a bacteria specific drug target.38
Enzymes Involved In Biosynthetic Pathway of Lysine
This review describes the essential details of the key
enzymes functioning in the lysine biosynthetic pathway
that are the products of essential bacterial genes that are
not expressed in humans. The pathway is of interest to
antibiotic discovery research. Accordingly, it also gives the
current status of rational drug design initiatives targeting
essential enzymes of the lysine biosynthesis pathway in
pathogenic bacteria. More recently, cloning and
expression of the DAP pathway components has
facilitated detailed investigations and structures of the
enzymes
have
been
determined
by
X-ray
crystallography.40
Dihydrodipicolinate
synthase
dihydrodipicolinate
synthase (DHDPS, EC 4.2.1.52, dapA)
DHDPS was first purified in 1965 from E.coli extracts and
isolated from the same bacteria, wheat, maize (Zea mays)
and higher plants. Kinetic studies suggest that pyruvate
(PYR) binds to the enzyme active site followed by loss of
water. The enzyme is the product of the dapA gene,
which has been shown to be essential in several bacterial
species.41 The currently accepted mechanism of DHDPS is
in which the structure of ASA is presumed to be the
hydrate (figure 4). In the first step of the mechanism, the
active site lysine (Lys161 in E.coli DHDPS) forms a Schiff
base with pyruvate, subsequent binding of the second
substrate ASA is followed by dehydration and cyclisation
to form the product.42,43 Based on the X-ray crystal
structure of the E.coli enzyme, sequence homologies with
DHDPS from other sources and site directed mutagenesis
studies, it is proposed that a catalytic triad of three
residues, tyrosine 133, threonine 44, and tyrosine 107
(E.coli numbering),44 act as a proton-relay to transfer
protons to and from the active site via a water-filled
channel leading to the lysine binding site and bulk
solvent. Subsequent binding and reaction of ASA then
takes place. Several approaches have been used to
investigate the enzyme active site. Initial studies showed
that a reducible imine (NaBH4 is inhibitory) is formed
between pyruvate and the α–amino group of a lysine
residue in the active site is confirmed by electrospray
mass spectrometry. Formation of an enamine at the
active site has been proven by enzyme catalysed
reversible exchange of tritium between β-H pyruvate and
45
water. In some organisms, the activity of DHDPS is
regulated allosterically by lysine via a classical feedback
inhibition process. Lysine feedback inhibition of DHDPS
has been investigated in several plants, gram-negative
46,47
and gram-positive bacterial species to date.
In
contrast, DHDPS from bacteria are significantly less
sensitive to lysine
counterparts.48
ISSN 0976 – 044X
inhibition
than
their
plant
Structure of DHDPS
The subunit and quaternary structure of DHDPS of E. coli ,
M. tuberculosis, N. meningitides, MRSA and several other
species is a homotetramer in both crystal structure and
solution (Figure 3).49 In E.coli, the monomer is 292 amino
acids in length and is composed of two domains. Each
monomer contains an N-terminal (β/α) 8-barrel (residues
1-224) with the active site located within the centre of
the barrel responsible for catalysis and regulation. The Cterminal domain (residues 225-292) consists of three αhelices and contains several key residues that mediate
tetramerisation.50 The association of the four monomers
leaves a large water-filled cavity in the centre of the
tetramer, The tetramer can also be described as a dimer
of dimers, with strong interactions between the
monomers A & B and C & D at the so-called tight dimer
interface and weaker interactions between the dimers AB and C-D at the weak dimer interface. All point
mutations resulted in destabilization of the ‘dimer of
dimers’ tetrameric structure. A tetrameric structure is not
essential for activity, confirming the importance of the
dimeric unit as the minimal functional assembly for
efficient lysine binding.51
Figure 3: The active sites, allosteric sites, dimerisation
interface (tight dimer interface) and tetramerisation
interface of E. coli DHDPS structure (PDB: 1YXC).
The active site is located in cavities formed by the two
monomers of the dimer. A long solvent-accessible
catalytic crevice with a depth of 10 Å is formed between
β-strands 4 and 5 of the barrel.52,53 Lys161, involved in
Schiff-base formation is situated in the β-barrel near the
catalytic triad of three residues, namely Tyr133, Thr44
and Tyr107, which act as a proton shuttle. Thr44 is
hydrogen bonded to both Tyr133 and Tyr10754 and its
position in the hydrogen-bonding network may play a role
in Schiff base formation and cyclisation. The dihedral
angles of Tyr107 fall in the disallowed region of the
Ramachandran plot. It involved in shuttling protons
between the active site and solvent. In contrast, Tyr133
plays an important role in substrate binding, donating a
proton to the Schiff base hydroxyl. It is also thought to
coordinate the attacking amino group of ASA, which
requires the loss of a proton subsequent to cyclisation
(Figure 4). A marked reduction in activity is observed in
International Journal of Pharmaceutical Sciences Review and Research
Available online at www.globalresearchonline.net
224
Int. J. Pharm. Sci. Rev. Res., 25(2), Mar – Apr 2014; Article No. 42, Pages: 221-230
single substitution mutants, highlighting the importance
of this catalytic triad.55
K161
NH2
H O
CO2
K161
Y133
H
H
O
O
O
Pyruvate
HO
H
HO
NH
O
HN
CO2
NH
O
H
CO2
O
G185
HTPA
CO2
H
HN
Y107
T44
O
H
CO2
NH2
O
H
O
H
HN
NH3
O
T44
K161
CO2
H2O
O H O
H
H
T44
O
O
HO
Y133
Solvent
H
HO
H
O
HN
NH3
Y107
T44
O
Y107
G185
G185
O2C
ASA
CO2
NH
Solvent
O
NH3
K161
Y133
H
HN
G185
O2C
O
NH2
O
Y107
O
Y107
T44
Cyclisation
Solvent
H
H
G185
Y133
H
Solvent
H
CO2
H
O2C
and undergo condensation with the enzyme, generating
an enzyme-bound adduct that closely resembles the
enzymatic intermediate. Inclusion of
hydroxyl
functionality in phenols 18b and 19b in order to better
mimic the enzymatic intermediate results in a switch of
enzyme inhibition mode to a slow-tight binding model.
Dihydrodipicolinate reductase (DHDPR, EC 1.3.1.26, dap
B)
Y133
O
Schiff Base Formation
K161
O H O
NH H
O
O
O
-H2O
-H
HO
NH
Solvent
H
O
Y107
T44
CO2
Y133
O
H
2
Solvent
H
O
K161
K161
Y133
H
O
Y107
T44
NH
G185
CO2
NH2
Solvent
O
Y107
T44
K161
Y133
H
CO2
HN
O
HO
NH2
ISSN 0976 – 044X
H
Figure 4: Proposed mechanism of DHDPS
Inhibitors of DHDPS
The design of inhibitors of DHDPS has traditionally been
based upon substrate and product analogy (figure 6).
Non-heterocyclic Analogues of pyruvate have shown
competitive inhibition with respect to pyruvate, and these
include 3-fluoropyruvate, ketobutyrate, ketovalerate,
glyoxylate and diketopimelic acid. Diketopimelic acid has
been reported to be an irreversible inhibitor of DHDPS.
The diene inhibitors, 4a and 4b, are more potent than the
corresponding mono-enes, 5a and 5b, with the diene
diester 4a being the best inhibitor of DHDPS in this series.
Mass spectrometric analyses have further explored the
enzyme–inhibitor interaction and determined the sites of
enzyme alkylation.56 The specificity of a range of
heterocyclic product analogues displayed clear
differentiation in inhibition of DHDPS enzymes from
different
pathogenic
species
like
B.anthracis,
M.tuberculosis and MRSA. This suggests that the
development of species-specific inhibitors of DHDPS as
potential targeted to specific pathogens.57 Product
analogues have been designed to mimic the heterocycles
DHDP and HTPA. A series of piperidine and pyridine-2,6dicarboxylate derivatives has been evaluated as potential
inhibitors of DHDPS. Amongst the heterocyclic analeugs
the piperidine diester 6b exhibited the most potent
inhibition of E.coli DHDPS. Chelidamic acid 8a displayed
high levels of inhibition of both DHDPS activity and
bacterial growth the planar compounds with 2,6substituents in a cis disposition, are more effective
inhibitors of DHDPS than the corresponding
transdisposed compounds (i.e., 7b and 10b are more
potent than 15b), Unlike DHDP, HTHDP an immediate
precursor of DHDP contains a hydroxyl group at the C-4
position. Therefore, compounds (Figure 6) were designed
as potential inhibitors containing oxygen functionality at
the C-4 position, thereby providing a lead in the
development of more potent inhibitors of DHDPS.58 Some
novel constrained bis(ketoacid) and bis(oximino-acid)
derivatives have been display time-dependent inhibition
DHDPR was first purified and isolated from E. coli in 1965.
Since further, the enzyme has been characterised from
several species including Bacillus spaericus, MRSA, M.
tuberculosis, S.aureus, and many other bacteria.59 In
MRSA, DHDPR is the product of the dapB gene. Analyses
of the dapB proteins from different bacterial species
suggest that two different classes of DHPR enzymes may
exist in bacteria. Like most dehydrogenases, DHDPR from
various bacterial species exhibits dual nucleotide
specificity. E. coli DHDPR and M. tuberculosis DHDPR have
been shown to have a preference for NADH over
NADPH.60 S.aureus DHDPR structure reveals different
conformational states of this enzyme even in the absence
of a substrate or nucleotide cofactor. The structure from
S. aureus provides a rationale-Lys35 compensates for the
co-factor site mutation. These observations are significant
for biligand inhibitor design that relies on ligand-induced
conformational changes as well as co-factor specificity for
this important drug target.61
Structure of DHDPR
The three-dimensional structure of DHDPR has been
elucidated by X-ray crystallography from different
bacterial species including, E. coli, M. tuberculosis and S.
aureus. DHDPR from E. coli was the first DHDPR enzyme
to be extensively studied in terms of structure and
function. The open reading frame codes for a 240 amino
acid protein with a monomeric molecular weight of
26,662 Da. DHDPR is a homotetramer with each
monomer unit consisting of an N-terminal nucleotide
binding (cofactor) domain and a C-terminal substrate
binding (tetramerization) domain linked by a flexible loop.
The active site is located at the interface between the
nucleotide binding and substrate binding domains (figure
5). Hydrogen exchange experiments and the crystal
structures of DHDPR bound to nucleotides and substrate
analogues suggest that the hydride transfer reaction
requires the movement of N-terminal domain towards
the C-terminal domain.61 The four monomeric subunits
interact with each other by forming a tetramerisation
interface consisting of a sixteen stranded central β-barrel
comprising 4 β-strands from each monomer.
The consensus sequence, E(L/A)HHXXKXDAPSGTA is
found in the substrate binding domain of all known
bacterial DHDPR enzymes. Molecular modelling studies,
using the apo form (enzyme in the absence of substrate)
of E. coli DHDPR as a structural template, suggest a
cluster of five basic residues are the key catalytic site
residues, namely His159, His160, Arg161, His162 and
Lys163 all contained within the consensus sequence.
International Journal of Pharmaceutical Sciences Review and Research
Available online at www.globalresearchonline.net
225
Int. J. Pharm. Sci. Rev. Res., 25(2), Mar – Apr 2014; Article No. 42, Pages: 221-230
These residues are located in the loop connecting βstrand B7 to α-helix A5. Structural studies of E. coli
DHDPR in complex with NADH and the substrate analogue
and inhibitor, 2,6-pyridinedicarboxylate (2,6-PDC), show
that 2,6-PDC is bound to the substrate binding domain of
DHDPR, in a spherical cavity bordered by residues from
both the nucleotide binding and substrate binding
domains (Figure 5). The bound inhibitor makes several
hydrogen bonding interactions with the atoms of the
conserved E(L/A)HHXXKXDAPSGTA
motif.
Similar
interactions are observed between 2,6-PDC and DHDPR
from M. tuberculosis.61
The nucleotide binding domain of DHDPR adopts a
Rossmann fold, which is typical of nucleotide-dependent
dehydrogenases (Figure 5). The consensus sequence
(V/I)(A/G)(V/I)-XGXXGXXG located within this domain, is
conserved in all NAD(P)H-dependent dehydrogenases,
including DHDPR. Structural analyses of E. coli DHDPR
show that this motif extends from the C-terminal end of
β-strand B1 to the loop that connects B1 to α-helix A1.
From the structures of DHDPR bound to NAD(P)H from E.
coli and M. tuberculosis it has been demonstrated that
the conserved nucleotide binding motif (GXXGXXG) and
the acidic residue (Glu38 in E. coli DHDPR) located 19-20
residues downstream of the glycine-rich region is
important in binding the cofactor.62 The two hydroxyl
groups from the adenine ribose are known to interact
with the side chain of Glu38 and also the backbone atoms
of the glycine rich motif GXXGXXG. Several hydrophobic
interactions exist between the adenine ring of NADH and
the residues Arg39, Gly84 and His88. The pyrophosphate
group of NADH is located over the α-helix A1 and
interacts with residues contained within the loop
connecting β-strand B1 and α-helix A1. MRSA–DHDPR
exhibits a unique nucleotide specificity utilizing NADPH as
a cofactor more effectively than NADH. Isothermal
titration calorimetry (ITC) studies reveal that MRSA–
DHDPR has 20-fold greater binding affinity for NADPH
relative to NADH. Enzyme follows a compulsory-order
ternary complex catalytic mechanism and cofactor
preference of MRSA–DHDPR and provides insight into
rational approaches to inhibiting this valid antimicrobial
62
target. .
ISSN 0976 – 044X
Inhibitors of DHDPR
DHDPR was screened for novel inhibitors by a molecular
modeling approach which used available crystal structure
of the enzyme with an inhibitor bound at active site
shown to be very effective in identifying novel inhibitors
of the enzyme as well as conventional screening proved
beneficial in identifying compounds with greater
structural diversity. Molecular modeling and X-ray crystal
structure of E. coli DHDP reductase with 2,6-PDC inhibitor
bound in active site was used with FLOG system for
database searching and docking to identify enzyme
inhibitors. Assignment of polar hydrogens, hydrogen
bonds and appropriate tautomeric states of imidazoles
from three dimensional structures typically shows that
each inhibitor candidate constriant 5 and 25
conformations, with heavy atoms represented by seven
atom types (hydrogen bond donors, hydrogen bond
acceptors, polar). A number of sulfonamide compounds
were found to be among most potent inhibitors
discovered through the FLOG-based molecular modeling
search. Heterocyclic and aromatic inhibitors of DHDPR
identified from general screening. All compounds are
competitive with respect to DHDP including The substrate
analogue, 2,6-PDC(21).63 Other substrate analogues such
as picolinic acid (22), isopthalic acid (23), pipecolic acid
(24) and dimethyl chelidamate (25), are much weaker
inhibitors. A vinylogous amide that acts as a competitive
inhibitor of DHDPR has been one of the most potent
inhibitors of it. Molecular modeling with conventional
drug screening strategies has identified novel inhibitors,
including sulfones and sulfonamides (Figure 6).
O
O
RO 2C
RO2C
CO2R
4a, R = Et
4b, R = H
RO2 C
N
H
CO2 R
6a
6b
5a, R = Et
5b, R = H
O
CO2R
RO2C
N
R=H
R = Me
7a
7b
O
S
CO2R
R=H
R = Me
O
S
RO2C
N
H
CO2R MeO2C
8a
8b
N
H
CO2Me ROOC
N
H
COOR
MeO2C
10a R= H
10b R= Me
9
R=H
R = Me
N
H
CO2Me
11
OH
O
S
ROOC
MeO2C
N
H
NC
CO2Me
N
CN MeO C
2
N
H
14
13
12
O
H
N
N
N
N MeO
N
N NH
N
N
16
N
NH
18a
18b
17
R'
NOH
NOH
COOR ROOC
ROOC
OMe
NH
15a R = H
15b R = Me
O
R'
COOR
N
H
CO2Me
R' = H
R' = OH
19a
19b
R = Me
R=H
COOR
R = Me
R = Me
R' = Me
R' = OH
Compounds 4-19 inhibiting DHDPS
NH2
O
S
O
O
H
N
HOOC
N
R1
R3
R2
23
O
20
O
Cl
R1 = CH3CH 2O
R1 = CH 3CH 2CH 2CH 2O
R1 = Ph
R1 = Ph
R1 = Ph
R2 = Cl R3 = NH2
R2 = H R3 = NH2
R2 = H R3 = NH2
R2 = Cl R3 = NH2
R2 = H R3 = CH 3CHCH3
N
H
COOH MeOOC
N
H
COOH HOOC
COOH
S
NH2
NH2
26
HOOC
COOH
NH2
OH
Figure 5: Structure of the E. coli DHDPR monomer bound
to NADH and the substrate analogue, 2,6-PDC (PDB:
1ARZ)
O
HOOC
COOH
S
S
O
27
COOMe
25
24
compounds 20-25 inhibiting DHDPR.
HOOC
NH2
COOH
22
O
S
O
20a
20b
20c
20d
20e
COOH HOOC
N
COOH
21
NH2
NH2
O
NH2
28
HOOC
COOH
HN
29
NH2
compounds 26-30 Inhibits DAPDC
30
NH2
Figure 6: Substrate based analogue of enzyme involve in
lysine biosynthesis.
International Journal of Pharmaceutical Sciences Review and Research
Available online at www.globalresearchonline.net
226
Int. J. Pharm. Sci. Rev. Res., 25(2), Mar – Apr 2014; Article No. 42, Pages: 221-230
Diaminopimelate dehydrogenase (DAPDH, EC 1.4.1.16) is
a NADPH dependant enzyme that catalyses the reductive
amination of L-2-amino-6-ketopimelate (AKP), the acyclic
form of L-2,3,4,5,-tetrahydrodipicolinate (THDP), to
produce meso-DAP (figure 2). It is assumed that the
reaction occurs via an imine intermediate as a result of
amination of AKP. Reduction of the imine by hydride
transfer from NADPH generates meso-DAP. Only a small
group of Gram-positive and Gram-negative bacteria
posses DAPDH activity. These include B. sphaericus,
Brevibacterium sp, C. glutamicum, Cl. thermocellum and
P. vulgaris.64 Characterised DAPDH enzymes are
comprised of approximately 320 residues and share
greater than 27% sequence identity across the species.
Some bacterial species possessing DAPDH activity use
multiple pathways to synthesise lysine. For example, C.
glutamicium can synthesise lysine by either the
dehydrogenase or succinylase pathway, whilst Bacillus
macerans can employ enzymes of the dehydrogenase or
acetylase pathways.65
Structure of DAPDH
DAPDH from C. glutamicum forms a homodimer of
approximately 70 kDa (Figure 7).65 The DAPDH monomer
subunit is comprised of three domain, a dinucleotide
binding domain, that is similar but not identical to
classical Rossman fold, a dimerisation domain, and a Cterminal domain (Figure 8). Monomer subunits interact
via two α-helices and three-stranded antiparallel β-sheet
to form dimer. Crystal structure of C. glutamicum DAPDH
in complex with ligand shows that oxidised cofactor,
NADP+, is bound within each of the dinucleotide binding
domains. Each monomer domains exhibit open and
closed conformations thought to represent binding and
active states of DAPDH. In closed conformation the
NADP+ pyrophosphate forms seven additional
noncovalent contacts. Subsequent studies demonstrate
the product, meso-DAP, binds within an elongated cavity
formed at interface of the dimerisation and dinucleotide
binding domains.66 In open conformer the dinucleotide is
accessible to solvent, while in closed conformer both
NADPH and DAP are protected from solvent. DAP binds in
an elongated cavity with hydrogen bond donors and
acceptors situated such that only D-amino acid center can
bind near the oxidized nucleotide. The carboxylate groups
of amino acid centers face cavity, bordered by protein
atoms, while amino groups are exposed to water.
ISSN 0976 – 044X
The dimerisation (orange), dinucleotide binding (blue),
and C-terminal (green) domains are indicated. The
cofactor NADPH (yellow) and inhibitor L-2-amino-6methylene-pimelate (yellow) are bound by active site
residues (pink) (PDB:1F06).
Inhibitors of DADPH
Crystal structures of C. glutamicum DAPDH complexes
with the inhibitors (2S,5S)-2-amino-3-(3-carboxy-2isoxazolin-5-yl)-propanoic
acid
and
L-2-amino-6methylene-pimelate show that they form similar
interactions with DAPDH as the product meso-DAP.67 The
structure of DAPDH with NADP+ and an isoxazoline
inhibitor bound was also solved. The inhibitor binds to the
DAP site, but with its L-amino acid center at the position
of the D-amino acid center of the substrate. This orients
the Cα-H bond away from the C4 position of the
nicotinamide ring of NADP+ and prevents catalysis.
6. Diaminopimelate Decarboxylase (DAPDC, EC 4. 1. 1.
20, lysA) DAPDC encoded by the lysA gene in bacteria,
which is an essential bacterial gene.68 In E.coli, lysine
specific gene lysA is transcriptionally controlled by the
LysR regulator protein.69 DAPDC is a vitamin B6dependent enzyme that catalyzing non-reversible
reaction in which DAPDC stereospecifically converts
meso-DAP to L-lysine and carbon dioxide. Unlike other
PLP-dependant decarboxylases that decarboxylate an Lstereocentre, DAPDC specifically cleaves the Dstereocentre carboxyl group. Thus, the enzyme possesses
a means to differentiate between two stereocentres.
DAPDC is classified as a type III class PLP enzyme, from
the alanine racemase family. The study on DAPDC
structure and function of this enzyme from H.pylori,
M.tubercolosis, and M.jannaschii. Structure of DAPDC
from M. tuberculosis have shown that the active site of
DAPDC is located at the dimer interface, the dimer is the
minimal catalytic requirment. In species such as M.
jannaschii and M. tuberculosis, DAPDC is function of a
homodimer and each subunits associate to form a headto-tail quaternary architecture (Figure 9).70
Figure 8: M. tuberculosis DAPDC monomer - The Nterminal (yellow) and C-terminal (magenta) domains are
indicated.
Structure of DAPDC
Figure 7: Structure of dimeric C. glutamicum DAPDH in
complex with NADPH and L-2-amino-6-methylenepimelate.
The DAPDC monomer is composed of two domains,
consisting of an N-terminal 8-fold α/β-barrel domain and
a C-terminal β-sheet domain (Figure 8). In M. tuberculosis
International Journal of Pharmaceutical Sciences Review and Research
Available online at www.globalresearchonline.net
227
Int. J. Pharm. Sci. Rev. Res., 25(2), Mar – Apr 2014; Article No. 42, Pages: 221-230
DAPDC, the N-terminal α/β-barrel domain (residues 48308) is comprised of β-strands β4–β13 and helices α2α10. The C-terminal domain (residues 2-47 and 309-446)
is comprised of β-strands β1-β3, β14-β21 and helices α1,
α11-α13. The active site is located at the interface
between the α/β-barrel domain of one subunit and βsheet domain of both subunits (Figure 8). The X-ray
structure of H. pylori DAPDC has allowed identification of
key residues involved in substrate and cofactor
recognition. The enzyme was crystallized in the presence
of PLP and lysine. The H. pylori structure is very similar to
that of M. tuberculosis DAPDC, forming a homodimer in a
head-to-tail conformation. In this enzyme, PLP forms
Schiff base linkages with Lys46 and lysine to produce a
lysine-PLP external aldimine. This aldimine is believed to
mimic the catalytic intermediate formed between mesoDAP and PLP.71,72
Inhibitors of DAPDC
Diaminopimelic acid analogues (Figure 6) described in the
section DAPE have been synthesised to study the
inhibition of DAPDC from different bacteria. Mixtures of
isomers of N-hydroxydiaminopimelate (29) and Naminodiaminopimelate (30) are potent competitive
inhibitors of DAPDC.73 Lanthionine sulfoxides (26 & 28)
are good competitive inhibitors with 50% inhibition at 1
mM. Weaker competitive inhibitors include the meso and
LL-isomers of lanthionine sulfone and lanthionine,
whereas the DD-isomers were less effective.74-76
3.
Simmons KJ, Chopra I, Fishwick WG, Structure-based discovery of
antibacterial drugs, nature reviews microbiology, 8, 2010, 501.
4.
Born TL, Blanchard JS, Structure /function studies on enzymes in
the diaminopimelate pathway of bacterial cell wall biosynthesis,
Chemical Biology, 3, 1999, 607–613.
5.
Saito Y, Shinkai T, Yoshimura Y, Takahata H, A straightforward
stereoselective
synthesis
of
meso-(S,S)-and(R,R)-2,6diaminopimelic acids from cis-1,4-diacetoxycyclohept-2-ene,
Bioorganic & Medicinal Chemistry Letters, 17, 2007, 5894–5896.
6.
Jurgens AR, Asymmetric Synthesis Of Differentially Protected
Meso-2,6-Diaminopimelic Acid, Tetrahedron Letters, 33, 1992,
4727-4730.
7.
Kimura K, Bugg TDH, Recent advances in antimicrobial nucleoside
antibiotics targeting cell wall biosynthesis, Natural Product Report,
20, 2003, 252–273.
8.
Girodeau JM, Agouridas C, Masson M, Pineau R, Goffic FL, The
Lysine Pathway as a Target for a New Genera of Synthetic
Antibacterial Antibiotics, Journal of Medicinal Chemistry, 29, 1986,
1023-1030.
9.
Collier PN, Patel I, Taylor RJK, concise A, stereoselective synthesis
of meso-2,6-diaminopimelic acid (DAP), Tetrahedron Letters, 42,
2001, 5953–5954.
10.
Rodionov DA, Vitreschak AG, Mironov AA, Gelfand MS, Regulation
of lysine biosynthesis and transport genes in bacteria: yet another
RNA riboswitch, Nucleic Acids Research, 31, 2003, 6748-6757.
11.
Paradisi F, Porzi G, Rinaldi S , Sandri S, A simple asymmetr ic
synthesis of (+)- and (-)-2,6-diaminopimelic acids, Tetrahedron:
Asymmetry, 11, 2000, 1259-1262.
12.
Winn M, Goss RJM, Kimura K, Bugg TDH, Antimicrobial nucleoside
antibiotics targeting cell wall assembly: Recent advances in
structure–function studies and nucleoside biosynthesis, Natural
Product Reports, 27, 2010, 279–304.
13.
Zoeiby A, Sanschagrin F, Levesque RC, Structure and function of
the Mur enzymes: development of novel inhibitors, Molecular
Microbiology, 47, 2003, 1–12.
14.
Cox RJ, The DAP Pathway to Lysine as a Target for Antimicrobial
Agents, natural product reports, 1996, 29-43.
15.
Hartmann M, Tauch A, Eggeling L, Bathe B, Identification and
characterization of the last two unknown genes, dapC and dapF, in
the succinylase branch of the L-lysine biosynthesis of
Corynebacterium glutamicum, Journal of Biotechnology, 104,
2003, 199-211.
16.
Galeazzi R, Garavelli M, Grandi A, Monari M, Porzi G, Sandri S,
Unusual peptides containing the 2,6-diaminopimelic acid
framework: Stereocontrolled synthesis, X-ray analysis, and
computational modelling. Part-2, Tetrahedron Asymmetry, 14,
2003, 2639–2649.
17.
Holcomb RC, Schow S, Kaloustian SA, Powell D, An Asymmetric
Synthesis Of Differentially Protected Meso-2,4-Diaminopimelic
Acid, Tetrahedron Letters, 35, 1994, 7005-7008.
18.
Chowdhury AR, Boons GJ, The synthesis of diaminopimelic acid
containing peptidoglycan fragments using metathesis cross
coupling, Tetrahedron Letters, 46, 2005, 1675–1678.
19.
Kosuge T, Hoshino T, Lysine is synthesized through the αaminoadipate pathway in Thermus thermophilus, FEMS
Microbiology Letters, 169, 1998, 361-367.
20.
Zabriskie TM, Jackson MD, Lysine biosynthesis and metabolism in
fungi, College of Pharmacy, Natural Product Report, 17, 2000, 85–
97.
21.
Nishida H, Nishiyama M, What Is Characteristic of Fungal Lysine
Synthesis Through the α-Aminoadipate Pathway, Journal of
Molecular Evolution, 51, 2000, 299–302.
CONCLUSION
The review thus focuses on the development of newer
chemical analogs acting as antimicrobials through
inhibition of enzymes especially involved in lysine
biosynthetic pathway. It imposes the need to develop
these analogs to fight with the resistance imposed by the
bacteria to the current antibiotics. Such compounds shall
be able to specifically target and inhibit enzymes and
proteins essential to the survival of bacteria. Selective
inhibition of the enzymes of DAP pathway by appropriate
substrate analogs might lead to newer drugs that are
more effective and less toxic to mammals. The drug
discovery towards protein and enzyme inhibition in this
way shall generate newer avenues in treatment of
diseases using antimicrobial therapy. Such drugs shall be
able to stand in the market for a longer duration of time
and also with a higher success rate.
Acknowledgements: The authors are thankful to the
Principal, Government College of Pharmacy, Aurangabad,
Maharashtra, India for providing facilities for literature
survey of this review article.
REFERENCES
1.
Barker J, Antibacterial drug discovery and structure based design,
Drug Discovery Today, 11, 2006, 9-10.
2.
Christopher W, Cloning of the dapB gene, encoding
dihydrodipicolinate reductase, from Mycobacterium tuberculosis,
Nature, 406, 2000, 17.
ISSN 0976 – 044X
International Journal of Pharmaceutical Sciences Review and Research
Available online at www.globalresearchonline.net
228
Int. J. Pharm. Sci. Rev. Res., 25(2), Mar – Apr 2014; Article No. 42, Pages: 221-230
22.
Kosuge T, Hoshino T, The α-Aminoadipate Pathway for Lysine
Biosynthesis Is Widely Distributed among Thermus Strains, Journal
Of Bioscience And Bioengineering, 88, 1999, 672-675.
23.
Miyazaki J, Kobashi N, Nishiyama M, Yamane H, Functional and
Evolutionary Relationship between Arginine Biosynthesis and
Prokaryotic Lysine Biosynthesis through α-Aminoadipate, Journal
of Bacteriology, 183, 2001, 5067.
24.
Johansson E, Steffens JJ, Lindqvist Y, Schneider G, Crystal Structure
of Saccharopine Reductase from Magnaporthe grisea, an Enzyme
of the α-Aminoadipate Pathway of Lysine Biosynthesis, Structure,
8, 2000, 1037–1047.
25.
Xu H, Andi B, Qian J, West AH, Cook PF, The α-Aminoadipate
Pathway for Lysine Biosynthesis in Fungi, Cell Biochemistry and
Biophysics, 46, 2006, 43-64.
26.
Nishida H, Nishiyama M, Kobashi N, A Prokaryotic Gene Cluster
Involved in Synthesis of Lysine through the Amino Adipate
Pathway: A Key to the Evolution of Amino Acid Biosynthesis,
Genome Research, 1999, 1175-1183.
27.
28.
29.
Ehmann DE, Gehring AM, Walsh CT, Lysine Biosynthesis in
Saccharomyces cerevisiae: Mechanism of R-Aminoadipate
Reductase
(Lys2)
Involves
Posttranslational
Phosphopantetheinylation by Lys5, Biochemistry, 38, 1999, 61716177.
Fazius F, Shelest E, Gebhardt P, Brock M, The fungal αaminoadipate pathway for lysine biosynthesis requires two
enzymes of the aconitase family for the isomerization of
homocitrate to homoisocitrate, Molecular Microbiology, 86, 2012,
1508–1530.
Horie H, Tomita T, Saiki A, Kono H, Taka H, Mineki R, Fujimura T,
Nishiyama C, Kuzuyama T, Nishiyama M, Discovery of
proteinaceous N-modification in lysine biosynthesis of Thermus
thermophilus, nature chemical biology, 5, 2009, 43–64.
30.
Fujiwara K, Tsubouchi T, Kuzuyama T, Nishiyama M, Involvement
of the arginine repressor in lysine biosynthesis of Thermus
thermophilus, Microbiology, 152, 2006, 3585–3594.
31.
Lende VD, Kamp MVD, Berg MVD, Klaas S, Bovenberg RAL,
Veenhuis M, γ-(L-α-Aminoadipyl)-L-cysteinyl-D-valine synthetase,
that mediates the first committed step in penicillin biosynthesis, is
a cytosolic enzyme, Fungal genetics and biology, 37, 2002, 49-55.
32.
Grundy FJ, Lehman SC, Henkin TM, The L box regulon: Lysine
sensing by leader RNAs of bacterial lysine biosynthesis genes,
National Academy of Sciences, 100, 2003, 12057–12062.
33.
Cahyanto MN, Kawasaki H, Nagashio M, Fujiyama K, Seki T,
Regulation
of
aspartokinase,
aspartate
semialdehyde
dehydrogenase,
Dihydrodipicolinate
synthase
and
dihydrodipicolinate reductase in Lactobacillus plantarum,
Microbiology, 152, 2006, 105–112.
34.
Tsujimoto N, Gunji Y, Miyata YO, Shimaoka M, Yasueda H, l-Lysine
biosynthetic pathway of Methylophilus methylotrophus and
construction of an l-lysine producer, Journal of Biotechnology, 124,
2006, 327–337.
35.
Born TL, Blanchard JS, Structure/function studies on enzymes in
the diaminopimelate pathway of bacterial cell wall biosynthesis,
Current Opinion in Chemical Biology, 3, 1999, 607–613.
36.
Dobson RCJ, Valegard K, Gerrard JA, The Crystal Structure of Three
Site-directed Mutants of Escherichia coli Dihydrodipicolinate
Synthase: Further Evidence for a Catalytic Triad, Journal of
Molecular, Biology, 338, 2004, 329–339.
37.
Rodionov DA, Vitreschak AG, Mironov AA, Gelfand MS, Regulation
of lysine biosynthesis and transport genes in bacteria: yet another
RNA riboswitch, Nucleic Acids Research, 31, 2003, 6748-6757.
38.
Dogovski C, Enzymology of Bacterial Lysine Biosynthesis,
Department of Biochemistry and Molecular Biology, Molecular
ISSN 0976 – 044X
Science and Biotechnology Institute, University of Melbourne,
Parkville, Victoria, Australia.
39.
Watanabe N, Cherney MM, Belkum MJ, Marcus SL, Flegel MD, Clay
MD, Deyholos MK, Vederas JC, James MNG, Crystal Structure of LLDiaminopimelate Aminomtransferase from Arabidopsis thaliana : A
Recently Discovered Enzyme in the Biosynthesis of L-Lysine by
Plants and Chlamydia, Journal of Molecular Biology, 371, 2007,
685– 702.
40.
Cox RJ, Sutherland A, Vederas JC, Bacterial Diaminopimelate
Metabolism as a Target for Antibiotic Design, Bioorganic &
Medicinal Chemistry, 8, 2000, 843-871.
41.
Kaur N, Gautam A, Kumar S, Singh A, Singh N, Sharma S, Sharma R,
Tewari R, Singh TP, Biochemical studies and crystal structure
determination of Dihydrodipicolinate synthase from Pseudomonas
aeruginosa, International Journal of Biological Macromolecules, 48,
2011, 779–787.
42.
Dobson RCJ, Griffin MDW, Roberts SJ, Gerrard JA,
Dihydrodipicolinate synthase (DHDPS) fromEscherichia coli displays
partial mixed inhibition with respect to its first substrate, pyruvate,
An International Journal of Biochemistry and Molecular Biology,
86, 2004, 311–315.
43.
Taylor ACM, Costa TPSD, Gerrard JA, New insights into the
mechanism of dihydrodipicolinate synthase using isothermal
titration calorimetry, An International Journal of Biochemistry and
Molecular Biology, 92, 2010, 254-262.
44.
Dobson RCJ, Valegard K, Gerrard JA, The Crystal Structure of Three
Site-directed Mutants of Escherichia coli Dihydrodipicolinate
Synthase: Further Evidence for a Catalytic Triad, Journal of
Molecular Biology, 338, 2004, 329–339.
45.
Evans G, Schuldt L, Griffin MDW, Devenish SRA, Pearce FG,
Perugini MA, Dobson RCJ, Jameson GB, Weiss MS, Gerrard JA, A
tetrameric structure is not essential for activity in
Dihydrodipicolinate synthase (DHDPS) from Mycobacterium
tuberculosis, Archives of Biochemistry and Biophysics, 512, 2011,
154–159.
46.
Girish TS, Sharma E, Gopal B, Structural and functional
characterization of Staphylococcus aureus dihydrodipicolinate
synthase, European Biochemical Societies Letters, 582, 2008,
2923–2930.
47.
Devenish SRA, Huisman FHA, Parker EJ, Hadfield AT, Gerrard JA,
Cloning and characterisation of dihydrodipicolinate synthase from
the pathogen Neisseria meningitides, Biochimica et Biophysica
Acta, 1794, 2009, 1168–1174.
48.
Blickling S, Beisel HG, Bozic D, Laber B, Huber R, Structure of
Dihydrodipicolinate Synthase of Nicotiana sylvestris Reveals Novel
Quaternary Structure, Journal Molecular Biology, 274, 1997, 608621.
49.
Griffin MDW, Dobson RCJ, Gerrard JA, Perugini MA, Exploring the
dihydrodipicolinate synthase tetramer: How resilientis the dimer–
dimer interface, Archives of Biochemistry and Biophysics, 494,
2010, 58–63.
50.
Guo BBB, Devenish SRA, Dobson RCJ, Taylor ACM, Gerrard JA, The
C-terminal domain of Escherichia coli dihydrodipicolinate synthase
(DHDPS) is essential for maintenance of quaternary structure and
efficient catalysis, Biochemical and Biophysical Research
Communications, 380, 2009, 802–806.
51.
Taylor ACM, Catchpole RJ, Dobson RCJ, Pearce FG, Perugini MA,
Gerrard JA, Disruption of quaternary structure in E. coli
Dihydrodipicolinate synthase (DHDPS) generates a functional
monomer that is no longer inhibited by lysine, Archives of
Biochemistry and Biophysics, 503, 2010, 202–206.
52.
Costa TPSD, Taylor ACM, Dobson RCJ, Devenish SRA, Jameson GB,
Gerrard JA, How essential is the ‘essential ’ active-site lysine in
dihydrodip icolinate synthase, Biochimie An International Journal
of Biochemistry and Molecular Biology, 92, 2010, 837-845.
International Journal of Pharmaceutical Sciences Review and Research
Available online at www.globalresearchonline.net
229
Int. J. Pharm. Sci. Rev. Res., 25(2), Mar – Apr 2014; Article No. 42, Pages: 221-230
53.
Mirwaldt C, Ingo K, Huber R, The Crystal Structure of
Dihydrodipicolinate Synthase from Escherichia coli at 2.5 Å
Resolution, Journal Molecular Biology, 246, 1995, 227–239.
54.
Kang BS, Kim YG, Ahn JW, Kim KJ, Crystal structure of
dihydrodipicolinate synthase from Hahella chejuensisat 1.5 Å
Resolution, International Journal of Biological Macromolecules, 46,
2010, 512–516.
55.
56.
57.
ISSN 0976 – 044X
fragilis and Clostridium thermocellum, Biochimica et Biophysica
Acta, 1814, 2011, 1162–1168.
66.
Wang F, Scapin G, Blanchard JS, Angelett RH, Substrate binding and
conformational
changes
of
Clostridium
glutamicum
diaminopimelate dehydrogenase revealed by hydrogen/deuterium
exchange and electrospray mass spectrometry, Protein Science,
1998, 7293-299.
Rice EA, Bannon GA, Glenn KC, Jeong SS, Sturman EJ, Rydel TJ,
Characterization and crystal structure of lysine insensitive
Corynebacterium glutamicum dihydrodipicolinate synthase
(cDHDPS) protein, Archives of Biochemistry and Biophysics, 480,
2008, 111–121.
67.
Nguyen L, Kozlov G, Gehring K, Structure of E. coli
tetrahydrodipicolinate Nsuccinyltransferase reveals the role of a
conserved C-terminal helix in cooperative substrate binding,
Federation of European Biochemical Societies Letters, 582, 2008,
623–626.
Boughton BA, Griffin MDW, O’Donnell PA, Dobson RCJ, Perugini
MA, Gerrard JA, Hutton CA, Irreversible inhibition of
dihydrodipicolinate synthase by 4-oxo-heptenedioic acid
analogues, Bioorganic & Medicinal Chemistry, 16, 2008, 9975–
9983.
68.
Saqib KM, Hay SM, Rees WD, The expression of Escherichia coil
diaminopimelate decarboxylase in mouse 3T3 cells, Biochimica et
Biophysica Acta, 1219, 1994, 398-404.
69.
Ikai H, Yamamoto S, Cloning and expression in Escherichia coli of
the gene encoding a novel L-2,4-diaminobutyrate decarboxylase of
Acinetobacter baumannii, FEMS Microbiology Letters, 124, 1994,
225-228.
70.
Gokulan K, Rupp B, Pavelka MS, Jacobs WR, Sacchettini JC, Crystal
Structure of Mycobacterium tuberculosisDiaminopimelate
Decarboxylase, an Essential Enzyme in Bacterial Lysine
Biosynthesis, The Journal Of Biological Chemistry, 278, 2003,
18588 –18596.
71.
Mills DA, Flickinger MC, Cloning and sequence analysis of the
meso-diaminopimelate decarboxylase gene from Bacillus
methanolicus MGA3 and comparison to other decarboxylase
genes, Applied and Environmental Microbiology, 59, 1993, 29272937.
Mitsakos V, Dobson RCJ, Pearce FG, Devenish SR, Evans GL,
Burgess BR, Perugini MA, Gerrard JA, Hutton CA, Inhibiting
dihydrodipicolinate synthase across species: Towards specificity
for pathogens, Bioorganic & Medicinal Chemistry Letters, 18, 2008,
842–844.
58.
Turner JJ, Gerrard JA, Hutton CA, Heterocyclic inhibitors of
dihydrodipicolinate synthase are not competitive, Bioorganic &
Medicinal Chemistry, 13, 2005, 2133–2140.
59.
Caplan JF, Zheng R, Blanchard JS, Vederas JC, Vinylogous Amide
Analogues of Diaminopimelic Acid (DAP) as Inhibitors of Enzymes
Involved in Bacterial Lysine Biosynthesis, Organic Letters, 2, 2000,
3857-3860.
60.
Dogovski C, Dommaraju SR, Small LC, Perugini MA, Comparative
structure and function analyses of native and his-tagged forms of
dihydrodipicolinate
reductase
from
methicillin-resistant
Staphylococcus aureus, Protein Expression and Purification, 85,
2012, 66–76.
72.
Ray SS, Bonanno JB, Rajashankar KR, Pinho MG, He G, Lencastre
HD, Tomasz A, Burley SK, Cocrystal Structures of Diaminopimelate
Decarboxylase: Mechanism, Evolution, and Inhibition of an
Antibiotic Resistance Accessory Factor, Structure, 10, 2002, 1499–
1508.
61.
Girish TS, Navratna V, Gopal B, Structure and nucleotide specificity
of Staphylococcus aureus Dihydrodipicolinate reductase (DapB),
Federation of European Biochemical Societies (FEBS) Letters, 585,
2011, 2561–2567.
73.
62.
Dommaraju SR, Dogovski C, Czabotar PE, Hor L, Smith BJ, Perugini
MA, Catalytic mechanism and cofactor preference of
dihydrodipicolinate
reductase
from
methicillin-resistant
Staphylococcus aureus, Archives of Biochemistry and Biophysics,
512, 2011, 167–174.
Lam LKP, Arnold LD, Kalantars TH, Kellands JG, Bell PML, Palcicg
MM, Pickardn MA, Vederas JC. Analogs of Diaminopimelic Acid as
Inhibitors of meso-Diaminopimelate Dehydrogenase and LLDiaminopimelate Epimerase, The Journal Biological Chemistry,
263, 1988, 11814-11819.
74.
Song Y, Niederer D, Bell PML, Lam LKP, Crawley S, Palcic MM,
Pickard MA, Pruess DL, Vederas JC, Stereospecific Synthesis of
Phosphonate Analogues of Diaminopimelic Acid (DAP), Their
Interaction with DAP Enzymes, and Antibacterial Activity of
Peptide Derivatives, Journal of Organic chemistry, 59, 1994, 57845793.
75.
Asschel IV, Soroka M, Haemersl A, Hooper M, Blanot D, Heijenoort
JV, Synthesis and antibacterial evaluation of phosphonic acid
analogues of diaminopimelic acid, European Journal of Medicinal
Chemistry, 26, 1991, 505-515.
76.
Liu H, Pattabiraman VR, Vederas JV, Stereoselective Syntheses of
4-Oxa Diaminopimelic Acid and Its Protected Derivatives via
Aziridine Ring Opening, Organic Letters, 9, 2007, 4211-4214.
63.
64.
65.
Paiva AM, Vanderwall DE, Blanchard JS, Kozarich JW, Williamson
JM, Kelly TM, Inhibitors of dihydrodipicolinate reductase, a key
enzyme of the diaminopimelate pathway of Mycobacterium
tuberculosis, Biochimica et Biophysica Acta, 1545, 2001, 67-77.
Akita H, Fujino Y, Doi K, hima TO, Highly stable mesodiaminopimelate
dehydrogenase
from
an
Ureibacillus
thermosphaericus strain A1 isolated from a Japanese compost:
purification, characterization and sequencing, AMB Express, 1,
2011, 1-43.
Hudson AO, Klartag A, Gilvarg C, Dobson RCJ, Marques FG, Leustek
T, Dual diaminopimelate biosynthesis pathways in Bacteroides
Source of Support: Nil, Conflict of Interest: None.
International Journal of Pharmaceutical Sciences Review and Research
Available online at www.globalresearchonline.net
230